Shared and Divergent Stimulatory Effects of S100A4 and Uric Acid on Proliferation and Chemotaxis of Mesenchymal Stem Cells (MSCs). Interplay Between Necrotic Tumor Material and Immunosuppressive MSCs within Tumor Microenvironment,

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 3239-3239
Author(s):  
Judith Luiza Eisenbacher ◽  
Tatjana Yildiz ◽  
Hubert Schrezenmeier ◽  
Ramin Lotfi

Abstract Abstract 3239 Background: Necrotic cell death is a characteristic feature of advanced solid tumor. S100 proteins and uric acid (UA) are released from necrotic (tumor) tissue regardless of tumor's origin. Both of these factors are known to influence immune response. Mesenchymal stem cells (MSCs) are often found within tumor microenvironment and are associated with poor prognosis of cancer patients in terms of metastasis and survival. MSCs seem to play a crucial role within tumor microenvironment probably due to their immunosuppressive capacity interfering with the specific anti-tumor immune response. Underlying mechanisms for MSC accumulation and stimulation in tumor tissue are not well characterized yet. S100A4 is known to promote metastasis in certain tumors such as colorectal cancer, but the exact mechanism of this effect still remains unclear. UA not only induces an acute inflammatory response but is also known to maintain chronic inflammation. In our previous experiments, we could already show that necrotic tumor material is capable of inducing chemotaxis and proliferation of MSCs. Focusing on S100A4 and UA, we now sought to characterize individual known factors within necrotic material which may be responsible for described effects. Materials and Methods: Proliferation assays were performed using CYQUANT Assay (Invitrogen) based on fluorescent staining of nucleic acid. Migration of fluorescent-labeled MSCs was assessed using FluoroBlok (BD) two-chamber chemotaxis plates, fluorescence within the bottom chamber of migration plates was measured with PolarStar plate reader (BMG). Results: We tested the influence of recombinant human S100A4 at concentrations between 50 and 1000 ng/ml on MSC proliferation and could demonstrate divergent effects depending on the chosen S100A4 concentration: S100A4 at concentrations not higher than 100 ng/ml enhanced the proliferation of MSCs in a dose dependent manner up to 50%, whereas concentrations above 100 ng/ml inhibited MSC proliferation down to 50% compared to medium containing 5% human serum without S100A4. We tested the chemotactic activity of recombinant human S100A4 at concentrations between 0.01 and 1000 ng/ml on MSCs and could demonstrate a dose dependent chemotactic activity up to 80% of the response which was achieved by positive control (50% FBS). By adding UA to suboptimal concentration of S100A4 (100ng/ml), we could enhance the chemotactic activity of S100A4 on MSCs. In contrast to its enhancing effect on MSC migration, UA inhibited dose dependently the proliferation inducing effect of S100A4 with strongest effect at 300μg UA/ml resulting in about 50% inhibition of proliferation compared to medium containing 100ng/ml S100A4 without additional UA. ELISA assays confirmed the presence of S100A4 in the necrotic tumor material which we had used in our previous experiments with similar stimulatory effects on MSCs in terms of proliferation and chemotaxis. Conclusions: MSCs are often found within tumor tissue and may influence the biologic behavior of tumor and host's immune response to tumor. We could characterize UA and S100A4 as bioactive factors within necrotic (tumor) material. Whereas S100A4 stimulated both proliferation as well as chemotaxis of MSCs, UA enhanced the chemotactic acitivity of S100A4 while inhibiting its proliferation inducing effect. Comparing our new data with our previous observations using tumor lysates to induce chemotaxis and proliferation of MSCs the overall effect of necrotic material seems to be rather proliferative and chemotactic even though some factors like UA may interfere with mentioned overall effects. Keeping in mind that MSCs can act as potent immunosuppressive cells, we conclude that tumor necrosis rather inhibits an effective anti-tumor immune response by favoring accumulation and proliferation of MSCs within tumor microenvironment. Our observations shed some light into the biology of MSCs within tumor microenvironment and open new questions concerning the interplay between MSCs, necrotic tumor cells and tumor progression. Possible strategies to break the described vicious circle by rather inducing an apoptotic tumor cell death or by reducing bioactive factors released from necrotic tissue within tumor microenvironment may be considered. Additionally, clinical investigations using specific antibodies to S100A4 or influencing serum concentrations of UA in tumor patients may be performed. Disclosures: No relevant conflicts of interest to declare.

2011 ◽  
Vol 41 (7) ◽  
pp. 2021-2028 ◽  
Author(s):  
Ramin Lotfi ◽  
Judith Eisenbacher ◽  
Ghasem Solgi ◽  
Karin Fuchs ◽  
Tatjana Yildiz ◽  
...  

2021 ◽  
Vol 21 (1) ◽  
Author(s):  
Surendar Aravindhan ◽  
Sura Salman Ejam ◽  
Methaq Hadi Lafta ◽  
Alexander Markov ◽  
Alexei Valerievich Yumashev ◽  
...  

AbstractA crosstalk established between tumor microenvironment and tumor cells leads to contribution or inhibition of tumor progression. Mesenchymal stem cells (MSCs) are critical cells that fundamentally participate in modulation of the tumor microenvironment, and have been reported to be able to regulate and determine the final destination of tumor cell. Conflicting functions have been attributed to the activity of MSCs in the tumor microenvironment; they can confer a tumorigenic or anti-tumor potential to the tumor cells. Nonetheless, MSCs have been associated with a potential to modulate the tumor microenvironment in favouring the suppression of cancer cells, and promising results have been reported from the preclinical as well as clinical studies. Among the favourable behaviours of MSCs, are releasing mediators (like exosomes) and their natural migrative potential to tumor sites, allowing efficient drug delivering and, thereby, efficient targeting of migrating tumor cells. Additionally, angiogenesis of tumor tissue has been characterized as a key feature of tumors for growth and metastasis. Upon introduction of first anti-angiogenic therapy by a monoclonal antibody, attentions have been drawn toward manipulation of angiogenesis as an attractive strategy for cancer therapy. After that, a wide effort has been put on improving the approaches for cancer therapy through interfering with tumor angiogenesis. In this article, we attempted to have an overview on recent findings with respect to promising potential of MSCs in cancer therapy and had emphasis on the implementing MSCs to improve them against the suppression of angiogenesis in tumor tissue, hence, impeding the tumor progression.


2016 ◽  
Vol 2016 ◽  
pp. 1-14 ◽  
Author(s):  
Drenka Trivanović ◽  
Jelena Krstić ◽  
Ivana Okić Djordjević ◽  
Slavko Mojsilović ◽  
Juan Francisco Santibanez ◽  
...  

State of tumor microenvironment (TME) is closely linked to regulation of tumor growth and progression affecting the final outcome, refractoriness, and relapse of disease. Interactions of tumor, immune, and mesenchymal stromal/stem cells (MSCs) have been recognized as crucial for understanding tumorigenesis. Due to their outstanding features, stem cell-like properties, capacity to regulate immune response, and dynamic functional phenotype dependent on microenvironmental stimuli, MSCs have been perceived as important players in TME. Signals provided by tumor-associated chronic inflammation educate MSCs to alter their phenotype and immunomodulatory potential in favor of tumor-biased state of MSCs. Adjustment of phenotype to TME and acquisition of tumor-promoting ability by MSCs help tumor cells in maintenance of permissive TME and suppression of antitumor immune response. Potential utilization of MSCs in treatment of tumor is based on their inherent ability to home tumor tissue that makes them suitable delivery vehicles for immune-stimulating factors and vectors for targeted antitumor therapy. Here, we review data regarding intrusive effects of inflammatory TME on MSCs capacity to affect tumor development through modification of their phenotype and interactions with immune system.


Author(s):  
Hariharan Jayaraman ◽  
Nalinkanth V. Ghone ◽  
Ranjith Kumaran R ◽  
Himanshu Dashora

: Mesenchymal stem cells because of its high proliferation, differentiation, regenerative capacity, and ease of availability have been a popular choice in cytotherapy. Mesenchymal Stem Cells (MSCs) have a natural tendency to home in a tumor microenvironment and acts against it, owing to the similarity of the latter to an injured tissue environment. Several studies have confirmed the recruitment of MSCs by tumor through various cytokine signaling that brings about phenotypic changes to cancer cells, thereby promoting migration, invasion, and adhesion of cancer cells. The contrasting results on MSCs as a tool for cancer cytotherapy may be due to the complex cell to cell interaction in the tumor microenvironment, which involves various cell types such as cancer cells, immune cells, endothelial cells, and cancer stem cells. Cell to cell communication can be simple or complex and it is transmitted through various cytokines among multiple cell phenotypes, mechano-elasticity of the extra-cellular matrix surrounding the cancer cells, and hypoxic environments. In this article, the role of the extra-cellular matrix proteins and soluble mediators that acts as communicators between mesenchymal stem cells and cancer cells has been reviewed specifically for breast cancer, as it is the leading member of cancer malignancies. The comprehensive information may be beneficial in finding a new combinatorial cytotherapeutic strategy using MSCs by exploiting the cross-talk between mesenchymal stem cells and cancer cells for treating breast cancer.


Molecules ◽  
2021 ◽  
Vol 26 (7) ◽  
pp. 1831
Author(s):  
Thitianan Kulsirirat ◽  
Sittisak Honsawek ◽  
Mariko Takeda-Morishita ◽  
Nuttanan Sinchaipanid ◽  
Wanvisa Udomsinprasert ◽  
...  

Andrographolide is a labdane diterpenoid herb, which is isolated from the leaves of Andrographis paniculata, and widely used for its potential medical properties. However, there are no reports on the effects of andrographolide on the human suprapatellar fat pad of osteoarthritis patients. In the present study, our goal was to evaluate the innovative effects of andrographolide on viability and Tri-lineage differentiation of human mesenchymal stem cells from suprapatellar fat pad tissues. The results revealed that andrographolide had no cytotoxic effects when the concentration was less than 12.5 µM. Interestingly, andrographolide had significantly enhanced, dose dependent, osteogenesis and chondrogenesis as evidenced by a significantly intensified stain for Alizarin Red S, Toluidine Blue and Alcian Blue. Moreover, andrographolide can upregulate the expression of genes related to osteogenic and chondrogenic differentiation, including Runx2, OPN, Sox9, and Aggrecan in mesenchymal stem cells from human suprapatellar fat pad tissues. In contrast, andrographolide suppressed adipogenic differentiation as evidenced by significantly diminished Oil Red O staining and expression levels for adipogenic-specific genes for PPAR-γ2 and LPL. These findings confirm that andrographolide can specifically enhance osteogenesis and chondrogenesis of mesenchymal stem cells from human suprapatellar fat pad tissues. It has potential as a therapeutic agent derived from natural sources for regenerative medicine.


2021 ◽  
Vol 19 (1) ◽  
Author(s):  
Jianjun Wu ◽  
Shoupin Xie ◽  
Hailong Li ◽  
Yanxia Zhang ◽  
Jia Yue ◽  
...  

Abstract Background Glioma is a complex cancer with a high morbidity and high mortality. Bone marrow mesenchymal stem cells (BMSCs) have shown promise as an excellent cell/drug delivery vehicle for gene-targeted therapy; however, maintaining genetic stability and biological activity remains difficult. Furthermore, whether BMSCs support or inhibit tumor growth remains debated. This study investigated whether a traditional Chinese medicine fomular, Fuzheng Yiliu decoction (FYD) had a synergistic antitumor effect with IL-12 gene-modified BMSCs in glioma-bearing nude mice Methods The lentivirus-mediated IL-12 gene was transfected into primarily cultured BMSCs. A total of 72 BALB/c nude mice were used to establish xenograft models with glioma U251 cells and were divided into groups (n = 12) including blank control group, nude mouse model group (model group), lentiviral transfection of BMSC group with no gene loading (BMSC group), IL-12 lentivirus-transfected BMSC group (IL-12 + BMSC group), FYD treatment group (FYD group), and FYD treatment in IL-12 lentivirus-transfected BMSC group (FYD + IL-12 + BMSC group).. After treatment for 14 days, all mice were sacrificed to collect tumor tissue and serum for more detection, such as distribution of BMSCs, cell apoptosis in xenograft tumors, serum IL-12 and INF-γ levels, mouse weight and tumor volume were measured Results There were significantly more apoptotic cells in tumor tissue in IL-12 gene transfected group, FYD treatment group and FYD combining with IL-12 gene transfected group than that in the model group (P < 0.05). The FYD + IL-12 + BMSC group showed significantly higher Bax and lower Bcl-2 expression (P < 0.05), and serum IL-12 and INF-γ levels (P < 0.05) were higher than that in all other groups. After the intervention, this group also showed a strong inhibitory effect against tumor growth (P < 0.05) Conclusions This study suggested FYD treatment combined with IL-12 gene-modified BMSCs shows synergistic antitumor effect in glioma-bearing nude mice.


2012 ◽  
Vol 21 (7) ◽  
pp. 1529-1545 ◽  
Author(s):  
Helena Motaln ◽  
Kristina Gruden ◽  
Matjaž Hren ◽  
Christian Schichor ◽  
Monika Primon ◽  
...  

2021 ◽  
Vol 22 ◽  
Author(s):  
Soheila Montazersaheb ◽  
Ezzatollah Fathi ◽  
Ayoub Mamandi ◽  
Raheleh Farahzadi ◽  
Hamid Reza Heidari

: Tumors are made up of different types of cancer cells that contribute to tumor heterogeneity. Among these cells, cancer stem cells (CSCs) have a significant role in the onset of cancer and development. Like other stem cells, CSCs are characterized by the capacity for differentiation and self-renewal. A specific population of CSCs is constituted by mesenchymal stem cells (MSCs) that differentiate into mesoderm-specific cells. The pro-or anti-tumorigenic potential of MSCs on the proliferation and development of tumor cells has been reported as contradictory results. Also, tumor progression is specified by the corresponding tumor cells like the tumor microenvironment. The tumor microenvironment consists of a network of reciprocal cell types such as endothelial cells, immune cells, MSCs, and fibroblasts as well as growth factors, chemokines, and cytokines. In this review, recent findings related to the tumor microenvironment and associated cell populations, homing of MSCs to tumor sites, and interaction of MSCs with tumor cells will be discussed.


Sign in / Sign up

Export Citation Format

Share Document